Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(47): e2303978120, 2023 Nov 21.
Article in English | MEDLINE | ID: mdl-37963252

ABSTRACT

Robust high-throughput assays are crucial for the effective functioning of a drug discovery pipeline. Herein, we report the development of Invasion-Block, an automated high-content screening platform for measuring invadopodia-mediated matrix degradation as a readout for the invasive capacity of cancer cells. Combined with Smoothen-Mask and Reveal, a custom-designed, automated image analysis pipeline, this platform allowed us to evaluate melanoma cell invasion capacity posttreatment with two libraries of compounds comprising 3840 U.S. Food and Drug Administration (FDA)-approved drugs with well-characterized safety and bioavailability profiles in humans as well as a kinase inhibitor library comprising 210 biologically active compounds. We found that Abl/Src, PKC, PI3K, and Ataxia-telangiectasia mutated (ATM) kinase inhibitors significantly reduced melanoma cell invadopodia formation and cell invasion. Abrogation of ATM expression in melanoma cells via CRISPR-mediated gene knockout reduced 3D invasion in vitro as well as spontaneous lymph node metastasis in vivo. Together, this study established a rapid screening assay coupled with a customized image-analysis pipeline for the identification of antimetastatic drugs. Our study implicates that ATM may serve as a potent therapeutic target for the treatment of melanoma cell spread in patients.


Subject(s)
Antineoplastic Agents , Ataxia Telangiectasia , Melanoma , Humans , Ataxia Telangiectasia/drug therapy , Cell Line, Tumor , Melanoma/drug therapy , Melanoma/metabolism , Antineoplastic Agents/pharmacology , High-Throughput Screening Assays , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism
2.
Pigment Cell Melanoma Res ; 33(6): 889-894, 2020 11.
Article in English | MEDLINE | ID: mdl-32511882

ABSTRACT

The role of the small GTPase RAB27A as an essential melanosome trafficking regulator in melanocytes is well-accepted. A decade ago, RAB27A was identified as a tumor dependency gene that promotes melanoma cell proliferation. RAB27A has since been linked to another propeller of cancer progression: exosome secretion. We have recently demonstrated that RAB27A is overexpressed in a subset of melanomas. High RAB27A gene and protein expression correlate with poor prognosis in melanoma patients. Mechanistic investigations revealed that the generation of pro-invasive exosomes was RAB27A-dependent and, therefore, silencing RAB27A reduced melanoma cell invasion in vitro and in vivo. However, previous studies have implicated RAB27A to be involved in both proliferation and invasion of melanoma cells. Employing four human cell lines, stratified by RAB27A expression, and one RAB27A-high mouse cell line, we demonstrate in this study that the effects of abrogating RAB27A expression on proliferation are only temporary, in contrast to our previously reported persistent effects on tumor invasion and metastasis. Therefore, we assist in the dissection of the short-term effects of RAB27A knockdown on melanoma cell proliferation versus long-term effects on melanoma invasion and metastasis. We believe that our findings provide novel insights into the effects of RAB27A blockade.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma/genetics , Melanoma/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , rab27 GTP-Binding Proteins/genetics , Cell Death , Cell Line, Tumor , Cell Proliferation/genetics , Gene Knockdown Techniques , Humans , rab27 GTP-Binding Proteins/metabolism
3.
Int J Cancer ; 144(12): 3070-3085, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30556600

ABSTRACT

Despite recent advances in targeted and immune-based therapies, advanced stage melanoma remains a clinical challenge with a poor prognosis. Understanding the genes and cellular processes that drive progression and metastasis is critical for identifying new therapeutic strategies. Here, we found that the GTPase RAB27A was overexpressed in a subset of melanomas, which correlated with poor patient survival. Loss of RAB27A expression in melanoma cell lines inhibited 3D spheroid invasion and cell motility in vitro, and spontaneous metastasis in vivo. The reduced invasion phenotype was rescued by RAB27A-replete exosomes, but not RAB27A-knockdown exosomes, indicating that RAB27A is responsible for the generation of pro-invasive exosomes. Furthermore, while RAB27A loss did not alter the number of exosomes secreted, it did change exosome size and altered the composition and abundance of exosomal proteins, some of which are known to regulate cancer cell movement. Our data suggest that RAB27A promotes the biogenesis of a distinct pro-invasive exosome population. These findings support RAB27A as a key cancer regulator, as well as a potential prognostic marker and therapeutic target in melanoma.


Subject(s)
Exosomes/metabolism , Melanoma/metabolism , Melanoma/pathology , rab27 GTP-Binding Proteins/metabolism , Animals , Cell Line, Tumor , Cell Movement/physiology , Clustered Regularly Interspaced Short Palindromic Repeats , Culture Media, Conditioned , Exosomes/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , Melanoma/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanosomes/genetics , Melanosomes/metabolism , Mice , Neoplasm Invasiveness , Nevus/genetics , Nevus/metabolism , Proteomics , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Spheroids, Cellular , rab27 GTP-Binding Proteins/biosynthesis , rab27 GTP-Binding Proteins/genetics
4.
Int J Cancer ; 142(9): 1926-1937, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29210065

ABSTRACT

Mutations in BRAF activate oncogenic MAPK signalling in almost half of cutaneous melanomas. Inhibitors of BRAF (BRAFi) and its target MEK are widely used to treat melanoma patients with BRAF mutations but unfortunately acquired resistance occurs in the majority of patients. Resistance results from mutations or non-genomic changes that either reactivate MAPK signalling or activate other pathways that provide alternate survival and growth signalling. Here, we show the histone deacetylase inhibitor (HDACi) panobinostat overcomes BRAFi resistance in melanoma, but this is dependent on the resistant cells showing a partial response to BRAFi treatment. Using patient- and in vivo-derived melanoma cell lines with acquired BRAFi resistance, we show that combined treatment with the BRAFi encorafenib and HDACi panobinostat in 2D and 3D culture systems synergistically induced caspase-dependent apoptotic cell death. Key changes induced by HDAC inhibition included decreased PI3K pathway activity associated with a reduction in the protein level of a number of receptor tyrosine kinases, and cell line dependent upregulation of pro-apoptotic BIM or NOXA together with reduced expression of anti-apoptotic proteins. Independent of these changes, panobinostat reduced c-Myc and pre-treatment of cells with siRNA against c-Myc reduced BRAFi/HDACi drug-induced cell death. These results suggest that a combination of HDAC and MAPK inhibitors may play a role in treatment of melanoma where the resistance mechanisms are due to activation of MAPK-independent pathways.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Melanoma/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Cell Line, Tumor , Drug Synergism , Heterografts , Histone Deacetylase Inhibitors/administration & dosage , Humans , Melanoma/enzymology , Mice , Mice, Inbred NOD , Mice, SCID , Protein Kinase Inhibitors/administration & dosage , Random Allocation , Signal Transduction/drug effects
5.
J Invest Dermatol ; 136(7): 1479-1489, 2016 07.
Article in English | MEDLINE | ID: mdl-26970356

ABSTRACT

The tumor microenvironment is characterized by cancer cell subpopulations with heterogeneous cell cycle profiles. For example, hypoxic tumor zones contain clusters of cancer cells that arrest in G1 phase. It is conceivable that neoplastic cells exhibit differential drug sensitivity based on their residence in specific cell cycle phases. In this study, we used two-dimensional and organotypic melanoma culture models in combination with fluorescent cell cycle indicators to investigate the effects of cell cycle phases on clinically used drugs. We demonstrate that G1-arrested melanoma cells, irrespective of the underlying cause mediating G1 arrest, are resistant to apoptosis induced by the proteasome inhibitor bortezomib or the alkylating agent temozolomide. In contrast, G1-arrested cells were more sensitive to mitogen-activated protein kinase pathway inhibitor-induced cell death. Of clinical relevance, pretreatment of melanoma cells with a mitogen-activated protein kinase pathway inhibitor, which induced G1 arrest, resulted in resistance to temozolomide or bortezomib. On the other hand, pretreatment with temozolomide, which induced G2 arrest, did not result in resistance to mitogen-activated protein kinase pathway inhibitors. In summary, we established a model to study the effects of the cell cycle on drug sensitivity. Cell cycle phase-specific drug resistance is an escape mechanism of melanoma cells that has implications on the choice and timing of drug combination therapies.


Subject(s)
Cell Cycle Checkpoints/drug effects , Drug Resistance, Neoplasm , Melanoma/metabolism , Skin Neoplasms/metabolism , Alkylating Agents/chemistry , Apoptosis , Bortezomib/chemistry , Cell Division , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dacarbazine/analogs & derivatives , Dacarbazine/chemistry , G1 Phase , G2 Phase , Humans , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Temozolomide
6.
J Biol Chem ; 290(15): 9588-603, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25720491

ABSTRACT

Multidrug resistance (MDR) is a major obstacle in cancer treatment. More than half of human cancers express multidrug-resistant P-glycoprotein (Pgp), which correlates with a poor prognosis. Intriguingly, through an unknown mechanism, some drugs have greater activity in drug-resistant tumor cells than their drug-sensitive counterparts. Herein, we investigate how the novel anti-tumor agent di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT) overcomes MDR. Four different cell types were utilized to evaluate the effect of Pgp-potentiated lysosomal targeting of drugs to overcome MDR. To assess the mechanism of how Dp44mT overcomes drug resistance, cellular studies utilized Pgp inhibitors, Pgp silencing, lysosomotropic agents, proliferation assays, immunoblotting, a Pgp-ATPase activity assay, radiolabeled drug uptake/efflux, a rhodamine 123 retention assay, lysosomal membrane permeability assessment, and DCF (2',7'-dichlorofluorescin) redox studies. Anti-tumor activity and selectivity of Dp44mT in Pgp-expressing, MDR cells versus drug-sensitive cells were studied using a BALB/c nu/nu xenograft mouse model. We demonstrate that Dp44mT is transported by the lysosomal Pgp drug pump, causing lysosomal targeting of Dp44mT and resulting in enhanced cytotoxicity in MDR cells. Lysosomal Pgp and pH were shown to be crucial for increasing Dp44mT-mediated lysosomal damage and subsequent cytotoxicity in drug-resistant cells, with Dp44mT being demonstrated to be a Pgp substrate. Indeed, Pgp-dependent lysosomal damage and cytotoxicity of Dp44mT were abrogated by Pgp inhibitors, Pgp silencing, or increasing lysosomal pH using lysosomotropic bases. In vivo, Dp44mT potently targeted chemotherapy-resistant human Pgp-expressing xenografted tumors relative to non-Pgp-expressing tumors in mice. This study highlights a novel Pgp hijacking strategy of the unique dipyridylthiosemicarbazone series of thiosemicarbazones that overcome MDR via utilization of lysosomal Pgp transport activity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Drug Resistance, Multiple/drug effects , Lysosomes/drug effects , Thiosemicarbazones/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antineoplastic Agents/pharmacology , Biological Transport/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Doxorubicin/pharmacology , Female , Humans , Lysosomes/metabolism , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Microscopy, Fluorescence , RNA Interference , Thiosemicarbazones/metabolism , Vinblastine/pharmacology , Xenograft Model Antitumor Assays
7.
Int J Cancer ; 135(5): 1060-71, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24531984

ABSTRACT

Amino acids, especially leucine and glutamine, are important for tumor cell growth, survival and metabolism. A range of different transporters deliver each specific amino acid into cells, some of which are increased in cancer. These amino acids consequently activate the mTORC1 pathway and drive cell cycle progression. The leucine transporter LAT1/4F2hc heterodimer assembles as part of a large complex with the glutamine transporter ASCT2 to transport amino acids. In this study, we show that the expression of LAT1 and ASCT2 is significantly increased in human melanoma samples and is present in both BRAF(WT) (C8161 and WM852) and BRAF(V600E) mutant (1205Lu and 451Lu) melanoma cell lines. While inhibition of LAT1 by BCH did not suppress melanoma cell growth, the ASCT2 inhibitor BenSer significantly reduced both leucine and glutamine transport in melanoma cells, leading to inhibition of mTORC1 signaling. Cell proliferation and cell cycle progression were significantly reduced in the presence of BenSer in melanoma cells in 2D and 3D cell culture. This included reduced expression of the cell cycle regulators CDK1 and UBE2C. The importance of ASCT2 expression in melanoma was confirmed by shRNA knockdown, which inhibited glutamine uptake, mTORC1 signaling and cell proliferation. Taken together, our study demonstrates that ASCT2-mediated glutamine transport is a potential therapeutic target for both BRAF(WT) and BRAF(V600E) melanoma.


Subject(s)
Amino Acid Transport System ASC/biosynthesis , Glutamine/metabolism , Large Neutral Amino Acid-Transporter 1/biosynthesis , Melanoma/pathology , Multiprotein Complexes/antagonists & inhibitors , Skin Neoplasms/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Amino Acid Transport System ASC/antagonists & inhibitors , Amino Acid Transport System ASC/genetics , Amino Acids, Cyclic/pharmacology , Benzyl Compounds/pharmacology , Biological Transport , CDC2 Protein Kinase/biosynthesis , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cell Survival , Humans , Leucine/metabolism , Mechanistic Target of Rapamycin Complex 1 , Melanoma/metabolism , Minor Histocompatibility Antigens , Multiprotein Complexes/genetics , Proto-Oncogene Proteins B-raf/genetics , RNA Interference , RNA, Small Interfering/genetics , Serine/analogs & derivatives , Serine/pharmacology , Signal Transduction , Skin Neoplasms/metabolism , Spheroids, Cellular , TOR Serine-Threonine Kinases/genetics , Tumor Cells, Cultured , Ubiquitin-Conjugating Enzymes/biosynthesis
8.
J Biol Chem ; 288(44): 31761-71, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24062304

ABSTRACT

Localization of the drug transporter P-glycoprotein (Pgp) to the plasma membrane is thought to be the only contributor of Pgp-mediated multidrug resistance (MDR). However, very little work has focused on the contribution of Pgp expressed in intracellular organelles to drug resistance. This investigation describes an additional mechanism for understanding how lysosomal Pgp contributes to MDR. These studies were performed using Pgp-expressing MDR cells and their non-resistant counterparts. Using confocal microscopy and lysosomal fractionation, we demonstrated that intracellular Pgp was localized to LAMP2-stained lysosomes. In Pgp-expressing cells, the Pgp substrate doxorubicin (DOX) became sequestered in LAMP2-stained lysosomes, but this was not observed in non-Pgp-expressing cells. Moreover, lysosomal Pgp was demonstrated to be functional because DOX accumulation in this organelle was prevented upon incubation with the established Pgp inhibitors valspodar or elacridar or by silencing Pgp expression with siRNA. Importantly, to elicit drug resistance via lysosomes, the cytotoxic chemotherapeutics (e.g. DOX, daunorubicin, or vinblastine) were required to be Pgp substrates and also ionized at lysosomal pH (pH 5), resulting in them being sequestered and trapped in lysosomes. This property was demonstrated using lysosomotropic weak bases (NH4Cl, chloroquine, or methylamine) that increased lysosomal pH and sensitized only Pgp-expressing cells to such cytotoxic drugs. Consequently, a lysosomal Pgp-mediated mechanism of MDR was not found for non-ionizable Pgp substrates (e.g. colchicine or paclitaxel) or ionizable non-Pgp substrates (e.g. cisplatin or carboplatin). Together, these studies reveal a new mechanism where Pgp-mediated lysosomal sequestration of chemotherapeutics leads to MDR that is amenable to therapeutic exploitation.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Lysosomal Membrane Proteins/metabolism , Lysosomes/metabolism , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration , Lysosomal-Associated Membrane Protein 2 , Lysosomal Membrane Proteins/genetics , Lysosomes/genetics , Neoplasm Proteins/genetics
9.
Mol Pharmacol ; 83(1): 179-90, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23074173

ABSTRACT

Deferasirox is an orally effective iron (Fe) chelator currently used for the treatment of iron-overload disease and has been implemented as an alternative to the gold standard chelator, desferrioxamine (DFO). Earlier studies demonstrated that DFO exhibits anticancer activity due to its ability to deplete cancer cells of iron. In this investigation, we examined the in vitro and in vivo activity of deferasirox against cells from human solid tumors. To date, there have been no studies to investigate the effect of deferasirox on these types of tumors in vivo. Deferasirox demonstrated similar activity at inhibiting proliferation of DMS-53 lung carcinoma and SK-N-MC neuroepithelioma cell lines compared with DFO. Furthermore, deferasirox was generally similar or slightly more effective than DFO at mobilizing cellular (59)Fe and inhibiting iron uptake from human transferrin depending on the cell type. However, deferasirox potently inhibited DMS-53 xenograft growth in nude mice when given by oral gavage, with no marked alterations in normal tissue histology. To understand the antitumor activity of deferasirox, we investigated its effect on the expression of molecules that play key roles in metastasis, cell cycle control, and apoptosis. We demonstrated that deferasirox increased expression of the metastasis suppressor protein N-myc downstream-regulated gene 1 and upregulated the cyclin-dependent kinase inhibitor p21(CIP1/WAF1) while decreasing cyclin D1 levels. Moreover, this agent increased the expression of apoptosis markers, including cleaved caspase-3 and cleaved poly(ADP-ribose) polymerase 1. Collectively, we demonstrate that deferasirox is an orally effective antitumor agent against solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoates/pharmacology , Iron Chelating Agents/pharmacology , Lung Neoplasms/drug therapy , Small Cell Lung Carcinoma/drug therapy , Triazoles/pharmacology , Administration, Oral , Animals , Antigens, CD/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Benzoates/therapeutic use , Cell Cycle/physiology , Cell Line, Tumor , Copper/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Deferasirox , Female , Humans , Iron/metabolism , Iron Chelating Agents/therapeutic use , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Neuroectodermal Tumors, Primitive, Peripheral , Protein Serine-Threonine Kinases/metabolism , Receptors, Transferrin/metabolism , Small Cell Lung Carcinoma/pathology , Transplantation, Heterologous , Triazoles/therapeutic use , Zinc/metabolism
10.
J Med Chem ; 55(16): 7230-44, 2012 Aug 23.
Article in English | MEDLINE | ID: mdl-22861499

ABSTRACT

We developed a series of second-generation di-2-pyridyl ketone thiosemicarbazone (DpT) and 2-benzoylpyridine thiosemicarbazone (BpT) ligands to improve the efficacy and safety profile of these potential antitumor agents. Two novel DpT analogues, Dp4e4mT and DpC, exhibited pronounced and selective activity against human lung cancer xenografts in vivo via the intravenous and oral routes. Importantly, these analogues did not induce the cardiotoxicity observed at high nonoptimal doses of the first-generation DpT analogue, Dp44mT. The Cu(II) complexes of these ligands exhibited potent antiproliferative activity having redox potentials in a range accessible to biological reductants. The activity of the copper complexes of Dp4e4mT and DpC against lung cancer cells was synergistic in combination with gemcitabine or cisplatin. It was demonstrated by EPR spectroscopy that dimeric copper compounds of the type [CuLCl](2), identified crystallographically, dissociate in solution to give monomeric 1:1 Cu:ligand complexes. These monomers represent the biologically active form of the complex.


Subject(s)
Antineoplastic Agents/chemical synthesis , Coordination Complexes/chemical synthesis , Copper , Ketones/chemical synthesis , Lung Neoplasms/drug therapy , Pyridines/chemical synthesis , Thiosemicarbazones/chemical synthesis , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Crystallography, X-Ray , Dimerization , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Injections, Intravenous , Ketones/chemistry , Ketones/pharmacology , Mice , Mice, Nude , Neoplasm Transplantation , Oxidation-Reduction , Pyridines/chemistry , Pyridines/pharmacology , Structure-Activity Relationship , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Transferrin/metabolism , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...